Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cell Proteomics ; 19(8): 1281-1302, 2020 08.
Article in English | MEDLINE | ID: mdl-32434922

ABSTRACT

Following central nervous system (CNS) demyelination, adult oligodendrocyte progenitor cells (OPCs) can differentiate into new myelin-forming oligodendrocytes in a regenerative process called remyelination. Although remyelination is very efficient in young adults, its efficiency declines progressively with ageing. Here we performed proteomic analysis of OPCs freshly isolated from the brains of neonate, young and aged female rats. Approximately 50% of the proteins are expressed at different levels in OPCs from neonates compared with their adult counterparts. The amount of myelin-associated proteins, and proteins associated with oxidative phosphorylation, inflammatory responses and actin cytoskeletal organization increased with age, whereas cholesterol-biosynthesis, transcription factors and cell cycle proteins decreased. Our experiments provide the first ageing OPC proteome, revealing the distinct features of OPCs at different ages. These studies provide new insights into why remyelination efficiency declines with ageing and potential roles for aged OPCs in other neurodegenerative diseases.


Subject(s)
Aging/metabolism , Oligodendrocyte Precursor Cells/metabolism , Proteome/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Separation , Cholesterol/metabolism , Myelin Sheath/metabolism , Neurodegenerative Diseases/pathology , Oligodendrocyte Precursor Cells/cytology , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Proteomics , Proteostasis , Rats, Sprague-Dawley , Reproducibility of Results
2.
Pain ; 159(3): 550-559, 2018 03.
Article in English | MEDLINE | ID: mdl-29351125

ABSTRACT

With less than 50% of patients responding to the current standard of care and poor efficacy and selectivity of current treatments, neuropathic pain continues to be an area of considerable unmet medical need. Biological therapeutics such as monoclonal antibodies (mAbs) provide better intrinsic selectivity; however, delivery to the central nervous system (CNS) remains a challenge. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is well described in inflammation-induced pain, and early-phase clinical trials evaluating its antagonism have exemplified its importance as a peripheral pain target. Here, we investigate the role of this cytokine in a murine model of traumatic nerve injury and show that deletion of the GM-CSF receptor or treatment with an antagonizing mAb alleviates pain. We also demonstrate enhanced analgesic efficacy using an engineered construct that has greater capacity to penetrate the CNS. Despite observing GM-CSF receptor expression in microglia and astrocytes, the gliosis response in the dorsal horn was not altered in nerve injured knockout mice compared with wild-type littermate controls as evaluated by ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein, respectively. Functional analysis of glial cells revealed that pretreatment with GM-CSF potentiated lipopolysaccharide-induced release of proinflammatory cytokines. In summary, our data indicate that GM-CSF is a proinflammatory cytokine that contributes to nociceptive signalling through driving spinal glial cell secretion of proinflammatory mediators. In addition, we report a successful approach to accessing CNS pain targets, providing promise for central compartment delivery of analgesics.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Analgesics/therapeutic use , Animals , Antibodies/therapeutic use , Brain/cytology , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Neuralgia/pathology , Neuroglia/drug effects , Signal Transduction/drug effects
3.
Neurosci Lett ; 545: 23-8, 2013 Jun 17.
Article in English | MEDLINE | ID: mdl-23603259

ABSTRACT

The expression of artemin (ARTN), a glial cell line-derived neurotrophic factor (GDNF) family ligand, increases in pre-clinical models of nociception and recent evidence suggests this growth factor may play a causative role in inflammatory pain mechanisms. The aim of this study was to demonstrate functional inhibition of ARTN with monoclonal antibodies and to determine whether ARTN neutralisation could reverse inflammatory pain in mice. We show that monoclonal antibodies with high affinity to ARTN, completely inhibit ARTN-induced Ret and ERK activation in a human neuroblastoma cell line, and block capsaicin-induced CGRP secretion from primary rat DRG cultures. In addition, administration of anti-ARTN antibodies to mice provides a transient, partial reversal (41%) of FCA-induced mechanical hypersensitivity. Anti-ARTN antibodies had no effect on hypersensitivity in response to partial nerve ligation in mice. These data suggest that ARTN-GFRα3 interactions partially mediate early stage nociceptive signalling following an inflammatory insult.


Subject(s)
Ganglia, Spinal/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hyperalgesia/physiopathology , Nerve Tissue Proteins/metabolism , Signal Transduction , Animals , Hot Temperature , Male , Protein Binding , Rats , Rats, Sprague-Dawley
4.
PLoS One ; 6(3): e14746, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21408068

ABSTRACT

BACKGROUND: Gap junction communication has been shown in glial and neuronal cells and it is thought they mediate inter- and intra-cellular communication. Connexin 36 (Cx36) is expressed extensively in the developing brain, with levels peaking at P14 after which its levels fall and its expression becomes entirely neuronal. These and other data have led to the hypothesis that Cx36 may direct neuronal coupling and neurogenesis during development. METHODOLOGY/PRINCIPAL FINDINGS: To investigate Cx36 function we used a neurosphere model of neuronal cell development and developed lentiviral Cx36 knockdown and overexpression strategies. Cx36 knockdown was confirmed by western blotting, immunocytochemistry and functionally by fluorescence recovery after photobleaching (FRAP). We found that knockdown of Cx36 in neurosphere neuronal precursors significantly reduced neuronal coupling and the number of differentiated neurons. Correspondingly, the lentiviral mediated overexpression of Cx36 significantly increased the number of neurons derived from the transduced neurospheres. The number of oligodendrocytes was also significantly increased following transduction with Cx36 indicating they may support neuronal differentiation. CONCLUSIONS/SIGNIFICANCE: Our data suggests that astrocytic and neuronal differentiation during development are governed by mechanisms that include the differential expression of Cx36.


Subject(s)
Cell Differentiation , Connexins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cell Aggregation , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Cells, Cultured , Connexins/genetics , Gap Junctions/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Vectors/genetics , In Situ Nick-End Labeling , Lentivirus/genetics , RNA, Small Interfering/metabolism , Rats , Reproducibility of Results , Gap Junction delta-2 Protein
5.
Mol Cell Neurosci ; 41(3): 373-82, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19332125

ABSTRACT

Inhibitors of the enzyme prolyl oligopeptidase (PO) improve performance in rodent learning and memory tasks. PO inhibitors are also implicated in the action of drugs used to treat bipolar disorder: they reverse the effects of three mood stabilizers on the dynamic behaviour of neuronal growth cones. PO cleaves prolyl bonds in short peptides, suggesting that neuropeptides might be its brain substrates. PO is located in the cytosol, however, where it would not contact neuropeptides. Here, we show that mice with a targeted PO null-mutation have altered growth cone dynamics. The wild-type phenotype is restored by PO cDNAs encoding either native or a catalytically-dead enzyme. In addition, we show that PO binds to the growth-associated protein GAP-43, which is a key regulator of synaptic plasticity. Taken together, our results show that peptidase activity is not required for PO function in neurons and suggest that PO instead acts by binding to cytosolic proteins that control growth cone and synaptic function.


Subject(s)
GAP-43 Protein/metabolism , Growth Cones/enzymology , Serine Endopeptidases/metabolism , Animals , Antimanic Agents/pharmacology , Carbamazepine/pharmacology , Cell Culture Techniques , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Growth Cones/drug effects , Humans , Indoles/pharmacology , Lamotrigine , Lithium Chloride/pharmacology , Mice , Mice, Knockout , Phosphatidylinositols/metabolism , Prolyl Oligopeptidases , Rats , Serine Endopeptidases/genetics , Thiazolidines/pharmacology , Triazines/pharmacology , Valproic Acid/pharmacology
6.
J Neurochem ; 108(4): 945-951, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19183265

ABSTRACT

Heat shock proteins (HSPs) are associated with the proteinaceous inclusions that characterise many neurodegenerative diseases. This suggests they may be associated with disease aetiology and/or represents an attempt to remove abnormal protein aggregates. In this study the adenoviral mediated over-expression of HSP70 interacting protein (HIP) alone was shown to significantly reduce inclusion formation in both an in vitro model of Spinal Bulbar Muscular Atrophy and a primary neuronal model of polyglutamine disease. Experiments to determine the mechanism of action showed that: denatured luciferase activity (a measure of protein refolding) was not increased in the presence of HIP alone but was increased when HIP was co-expressed with HSP70 or Heat Shock cognate protein 70 (HSC70); the expression of polyglutamine inclusions in cortical neurons mediated an increase in the levels of HSC70 but not HSP70. Our data suggest that HIP may prevent inclusion formation by facilitating the constitutive HSC70 refolding cycle and possibly by preventing aggregation. HIP expression is not increased following stress and its over-expression may therefore reduce toxic polyglutamine aggregation events and contribute to an effective therapeutic strategy.


Subject(s)
Carrier Proteins/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Inclusion Bodies/metabolism , Muscular Atrophy, Spinal/metabolism , Neurons/metabolism , Peptides/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Genetic Predisposition to Disease/genetics , Genetic Vectors , HSC70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/physiopathology , Humans , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Mice , Models, Biological , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Neurons/pathology , Peptides/genetics , Protein Folding , Rats , Rats, Wistar , Transfection
7.
Neurosci Lett ; 448(1): 71-3, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18950680

ABSTRACT

Multiple sclerosis is a currently incurable inflammatory demyelinating syndrome. Recent reports suggest that bone marrow derived mesenchymal stem cells may have therapeutic potential in experimental models of demyelinating disease, but various alternative mechanisms, ranging from systemic immune effects to local cell replacement, have been proposed. Here we used intraperitoneal delivery of human mesenchymal stem cells to help test (a) whether human cells can indeed suppress disease, and (b) whether CNS infiltration is required for any beneficial effect. We found pronounced amelioration of clinical disease but profoundly little CNS infiltration. Our findings therefore help confirm the therapeutic potential of mesenchymal stem cells, show that this does indeed extend to human cells, and are consistent with a peripheral or systemic immune effect of human MSCs in this model.


Subject(s)
Cell Transplantation/physiology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Antigens, Nuclear/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Glycoproteins/administration & dosage , Green Fluorescent Proteins/genetics , Humans , Injections, Intraperitoneal/methods , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Severity of Illness Index , Transfection/methods , Transplantation, Heterologous/methods
8.
Arterioscler Thromb Vasc Biol ; 28(8): 1462-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18497305

ABSTRACT

BACKGROUND: Generation of physiologically active vascular beds by delivery of combinations of growth factors offers promise for vascular gene therapy. METHODS AND RESULTS: In a mesenteric model of physiological angiogenesis, combining endothelial nitric oxide synthase (eNOS) (and hence NO production) with VEGF and angiopoietin-1 overexpression resulted in a more functional vascular phenotype than growth factor administration alone. eNOS gene delivery upregulated eNOS, VEGF, and Ang-1 to similar levels as gene transfer with VEGF or Ang-1. eNOS overexpression resulted in neovascularization to a similar extent as VEGF and Ang-1 combined, but not by sprouting angiogenesis. Whereas combining Ang-1 and VEGF increased both exchange vessels and conduit vessels, neither growth factor nor eNOS alone resulted in vessels with smooth muscle cell (SMC) coverage. In contrast, combining all three generated microvessels with SMCs (arteriolar genesis) and further increased functional vessels. Use of a vasodilator, prazosin, in combination with Ang1 and VEGF, but not alone, also generated SMC-positive vessels. CONCLUSIONS: Coexpression of eNOS, VEGF, and Ang-1 results in a more mature vascularization of connective tissue, and generates new arterioles as well as new capillaries, and provides a more physiological therapeutic approach than single growth factor administration, by combining hemodynamic forces with growth factors.


Subject(s)
Neovascularization, Physiologic/physiology , Splanchnic Circulation/physiology , Angiopoietin-1/physiology , Animals , Male , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Nitric Oxide Synthase Type II/physiology , Nitric Oxide Synthase Type III , Pericytes/physiology , Rats , Regional Blood Flow/physiology , Vascular Endothelial Growth Factor A/physiology
9.
J Neuroimmunol ; 193(1-2): 59-67, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18061686

ABSTRACT

Mesenchymal stem cells (MSCs) have the potential to play a role in autologous treatment of central nervous system injury or disease. Here we transduced human MSCs with enhanced green fluorescent protein (EGFP). We compared the capacity of control and EGFP-positive cells to proliferate under normal culture conditions, as well as express neural markers following trans-differentiation. EGFP-positive cells proliferated comparably to controls, retained EGFP expression over the course of multiple passages, and retained neural marker expression at levels comparable to control MSCs. Further neurogenic capacity of EGFP-positive human MSCs was examined by growth as neural stem cell-like neurospheres. No significant difference was observed in the ability of control or EGFP-positive cells to generate primary neurospheres or to expand during passage. When examined by immunostaining for the presence of neuroectodermal markers, neurosphere-derived cells similarly expressed neural markers. We show that human MSCs expressing EGFP represent an attractive and practical source of stem cells for the study of repair and regeneration in neurological models.


Subject(s)
Cell Differentiation , Green Fluorescent Proteins/genetics , Mesenchymal Stem Cells/cytology , Neurons/cytology , Biomarkers , Cell Proliferation , Cells, Cultured , Central Nervous System Diseases/therapy , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Transduction, Genetic
10.
Microcirculation ; 13(6): 423-37, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16864410

ABSTRACT

OBJECTIVE: Therapeutic angiogenesis requires an understanding of how growth factors such as vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) result in physiological neovascularization. This study determined the physiological mechanism by which adenoviral delivery of growth factor combinations alter vascular phenotype and functionality. METHODS: Adenovirus-mediated gene transfer into the adjacent fat pad of the rat mesentery was used to characterize induction of angiogenesis by VEGF and Ang-1, in a model that permitted a detailed examination of the neovessel phenotype. RESULTS: Ang-1 combined with VEGF resulted in a distinct vascular phenotype from either factor alone. Microvascular perfusion was significantly enhanced in all groups, but VEGF produced short, narrow, highly branched and sprouting vessels, with normal pericyte coverage. Ang-1 induced broader, longer neovessels, with no increase in branching or sprouting, yet a significantly higher pericyte ensheathment. Combination of Ang-1 and VEGF generated a significantly higher degree of functionally perfused, larger, less branched, and more mature microvessels, resulting from increased efficiency of sprout to vessel formation. Ang-1 and VEGF also caused differential effects on larger compared with smaller blood vessels, a finding reproduced in vitro. CONCLUSIONS: Ang-1 and VEGF use different physiological mechanisms to enhance neovascularization of relatively avascular tissue. Administration of both growth factors combines these physiological mechanisms to give greater enhancement of neovascularization than either growth factor alone. These results suggest that effective revascularization therapy may require combination growth factor treatment.


Subject(s)
Adenoviridae , Angiopoietin-1/biosynthesis , Mesentery/blood supply , Neovascularization, Physiologic/physiology , Transduction, Genetic , Vascular Endothelial Growth Factor A/biosynthesis , Angiopoietin-1/genetics , Animals , Humans , Male , Mesentery/cytology , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/genetics
11.
J Neurosci ; 25(27): 6296-303, 2005 Jul 06.
Article in English | MEDLINE | ID: mdl-16000619

ABSTRACT

We established the importance of phosphorylation of cAMP responsive element-binding protein (CREB) to both the familiarity discrimination component of long-term recognition memory and plasticity within the perirhinal cortex of the temporal lobe. Adenoviral transduction of perirhinal cortex (and adjacent visual association cortex) with a dominant-negative inhibitor of CREB impaired the preferential exploration of novel over familiar objects at a long (24 h) but not a short (15 min) delay, disrupted the normal reduced activation of perirhinal neurons to familiar compared with novel pictures, and impaired long-term potentiation of synaptic transmission in perirhinal slices. The consistency of these effects across the behavioral, systems, and cellular levels of analysis provides strong evidence for involvement of CREB phosphorylation in synaptic plastic processes within perirhinal cortex necessary for long-term recognition memory.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Exploratory Behavior/physiology , Long-Term Potentiation/physiology , Pattern Recognition, Visual/physiology , Protein Processing, Post-Translational , Temporal Lobe/physiology , Adenoviridae/genetics , Animals , Association Learning , Cyclic AMP/physiology , Defective Viruses/genetics , Discrimination Learning/physiology , Discrimination, Psychological/physiology , Entorhinal Cortex/physiology , Excitatory Postsynaptic Potentials , Genes, Reporter , Genes, fos , Genetic Vectors/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , In Vitro Techniques , Male , Phosphorylation , Photic Stimulation , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Recombinant Fusion Proteins/genetics , Single-Blind Method , Synaptic Transmission/physiology , Transcription, Genetic , Transduction, Genetic
12.
J Neurosci Res ; 79(5): 661-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15657876

ABSTRACT

Antisense technology, including ribozyme and small interfering RNA, is being developed to mediate the down-regulation of specific intracellular genes. It was observed in this study that both antiluciferase ribozymes and short hairpin RNAs (shRNAs) could significantly reduce the activity of exogenously expressed luciferase in primary hippocampal neurons in a viral titer-dependent manner. shRNAs were more effective gene-silencing agents than ribozymes, although they exhibited some nonspecific gene-silencing effects at high viral titers. We also attempted to increase ribozyme efficacy by using a woodchuck hepatitis posttranscriptional regulatory element (WPRE) in the ribozyme expression cassette. The results showed that adenoviral vectors encoding specific ribozymes could silence the cellular expression of luciferase and endogenous procaspase-3 significantly. Furthermore, the antiprocaspase-3 ribozyme was shown to inhibit staurosporine-mediated cell death. The addition of a WPRE did not, however, increase or decrease ribozyme activity. As far as we are aware, this is the first example of adenovirally mediated delivery of hammerhead ribozymes being used to manipulate gene expression in primary neurons. The results therefore suggest that hammerhead ribozymes may be useful tools for studying neuronal gene function and have potential as therapeutic agents to treat CNS diseases.


Subject(s)
Adenoviridae/physiology , Apoptosis/drug effects , Caspase Inhibitors , Neurons/metabolism , RNA, Catalytic/pharmacology , Animals , Apoptosis/physiology , Benzimidazoles/metabolism , Blotting, Western/methods , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cloning, Molecular , Embryo, Mammalian , Genetic Vectors/physiology , Hippocampus/cytology , Humans , Luciferases/metabolism , Neurons/virology , RNA, Messenger/biosynthesis , RNA, Viral/metabolism , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transduction, Genetic/methods
13.
Exp Physiol ; 90(1): 33-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15542617

ABSTRACT

Inducible gene expression systems have typically encountered limitations, such as pleitropic effects of the inducer, basal leakiness, toxicity of inducing agents and low levels of expression. However, recently non-toxic, tightly regulated control of transgene expression has been reported for several systems, the most frequently cited being the tetracycline gene control system. We have found that the individual components of the Tet system [the Tet transactivators and tetracycline responsive element (TRE)] function optimally to control gene expression when they are incorporated into separate adenoviral vectors. Furthermore, incorporation of the Woodchuck hepatitis virus post-transcriptional enhancer (WPRE) allows a dual vector Tet-regulatable Ad system to be used at very low titres (2 x 10(4)) that elicit a minimal inflammatory response, with no loss of transgene expression or ability to regulate transgene expression. This and similar regulatable systems will benefit studies investigating neuronal gene function and those seeking to develop effective neuronal gene therapy strategies.


Subject(s)
Adenoviridae/genetics , DNA, Viral/administration & dosage , DNA, Viral/genetics , Gene Expression Regulation/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Genetic Vectors/genetics , Animals , Genetic Therapy/methods , Humans , Tetracycline
14.
J Gene Med ; 7(5): 576-83, 2005 May.
Article in English | MEDLINE | ID: mdl-15580589

ABSTRACT

BACKGROUND: In previous studies we have found that the tetracycline (Tet)-regulatable system functions best in recombinant adenoviral (Ad) vectors when the Tet transactivators and the Tet-regulatable element (TRE) are incorporated into separate viral vectors. However, such a dual vector system is disadvantaged by the need to use relatively high titres that may elicit an immune response. Therefore, to develop a system that could be used at low titres while mediating strong, tightly regulatable gene expression in the central nervous system (CNS), we incorporated the woodchuck hepatitis virus post-transcriptional enhancer (WPRE) into a neuron-specific Tet-regulatable Ad system. METHODS: The WPRE was incorporated into Ad vectors encoding the Tet-Off (tTA) transactivator driven by the synapsin-1 and CMV promoters and encoding the TRE driving EGFP expression (TRE)-EGFP. RESULTS: The addition of the WPRE to the neuron-specific Tet-regulatable system mediated a greater than three-fold increase in transgene expression in primary hippocampal neurons with no loss of gene regulation. The results also showed that the addition of the WPRE enhanced transgene expression in the CNS without the loss of neuron specificity and without affecting the ability to regulate transgene expression. CONCLUSIONS: We have further developed a tetracycline-regulatable neuron-specific expression system such that it can now be used at low titres with no loss of transgene expression or ability to regulate transgene expression. It should therefore be of significant value to studies investigating neuronal gene function and to those seeking to develop effective neuronal gene therapy strategies.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/administration & dosage , Hippocampus/metabolism , Neurons/metabolism , Synapsins/genetics , Synapsins/metabolism , Tetracycline/pharmacology , Animals , Enhancer Elements, Genetic , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Hepatitis B Virus, Woodchuck/genetics , Hippocampus/embryology , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Wistar , Regulatory Sequences, Nucleic Acid , Transduction, Genetic , Transfection , Transgenes/physiology
15.
Neuroreport ; 15(7): 1171-5, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15129168

ABSTRACT

In this study we have used a molecular approach to manipulate CREB gene expression to study its role in the regulation of neuronal cell death. To achieve this, adenoviral (Ad) vectors encoding EGFP, CREB, and a powerful CREB dominant-negative, known as A-CREB were constructed. The over-expression of CREB but not A-CREB was found to protect primary hippocampal neurons from staurosporine-induced apoptosis, glutamate induced excitotoxicity and exposure to an in vitro ischaemic stress. Hence, manipulating CREB-regulated pathways may provide a means of delaying or preventing the neuronal cell death associated with ischaemic related injury, and in neurodegenerative diseases such as Huntington's and Alzheimer's disease.


Subject(s)
Apoptosis/physiology , Cyclic AMP Response Element-Binding Protein/biosynthesis , Excitatory Amino Acid Agonists/pharmacology , Neurons/metabolism , Stress, Physiological/metabolism , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HeLa Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Neurons/drug effects , Rats , Rats, Wistar , Stress, Physiological/genetics
16.
J Gene Med ; 5(7): 554-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12825194

ABSTRACT

BACKGROUND: Adenoviral (Ad) vectors are one of the most widely used tools for modelling gene therapy strategies. However, they have not been used in long-term models of neurological disease, as the period of time for which they mediate strong transgene expression is limited and/or variable. In this study we investigated the longevity of transgene expression in the brain when the powerful neuron-specific Ad-synapsin (Sy)-EGFP-woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) vector cassette is used at titres that do not elicit an immune response. METHODS: Adenoviral vectors expressing enhanced green fluorescent protein (EGFP) under the control of either the hCMV, hCMV-WPRE, Sy or Sy-WPRE promoter were constructed. These vectors were injected into the dentate gyrus region of hippocampus and transgene expression and immune cell infiltration assessed by fluorescence microscopy and immunocytochemical techniques, respectively. RESULTS: The quantitative analysis of EGFP expression showed that there was no significant change in synapsin or synapsin-WPRE driven transcription 9 months after injection when compared with expression levels obtained 3 days after injection. However, when the hCMV promoter or the hCMV-WPRE promoter cassette drove transgene expression, there was a dramatic fall in expression levels and very little expression was seen 9 months post-transfection. CONCLUSIONS: This study shows that non-integrating vectors can be used to mediate powerful, long-term episomal transgene expression in neurones. This work has important implications for neuronal gene therapy and is of relevance to studies investigating memory, behaviour and neuronal gene function.


Subject(s)
Adenoviridae/genetics , Brain/metabolism , Genetic Vectors , Hippocampus/metabolism , Neurons/metabolism , Promoter Regions, Genetic , Synapsins/genetics , Animals , Brain/virology , Cell Line , Gene Expression , Gene Transfer Techniques , Green Fluorescent Proteins , Hepatitis B Virus, Woodchuck/genetics , Humans , Luminescent Proteins , Male , Neurons/virology , RNA Processing, Post-Transcriptional , Rats , Rats, Wistar , Synapsins/metabolism , Transgenes
17.
Mol Ther ; 7(5 Pt 1): 588-96, 2003 May.
Article in English | MEDLINE | ID: mdl-12718901

ABSTRACT

Due to the complexity of brain function and the difficulty in monitoring alterations in neuronal gene expression, the potential of lentiviral gene therapy vectors to treat disorders of the CNS has been difficult to fully assess. In this study, we have assessed the utility of a third-generation equine infectious anemia virus (EIAV) in the Brattleboro rat model of diabetes insipidus, in which a mutation in the arginine vasopressin (AVP) gene results in the production of nonfunctional mutant AVP precursor protein. Importantly, by using this model it is possible to monitor the success of the gene therapy treatment by noninvasive assays. Injection of an EIAV-CMV-AVP vector into the supraoptic nuclei of the hypothalamus resulted in expression of functional AVP peptide in magnocellular neurons. This was accompanied by a 100% recovery in water homeostasis as assessed by daily water intake, urine production, and urine osmolality lasting for a 1-year measurement period. These data show that a single gene defect leading to a neurological disorder can be corrected with a lentiviral-based strategy. This study highlights the potential of using viral gene therapy for the long-term treatment of disorders of the CNS.


Subject(s)
Arginine Vasopressin/genetics , Diabetes Insipidus, Neurogenic/therapy , Genetic Therapy , Genetic Vectors , Infectious Anemia Virus, Equine/genetics , Supraoptic Nucleus/metabolism , Animals , Arginine Vasopressin/metabolism , Diabetes Insipidus, Neurogenic/metabolism , Diabetes Insipidus, Neurogenic/pathology , Homeostasis , Humans , In Situ Hybridization , Male , Rats , Rats, Brattleboro , Rats, Inbred WKY , Supraoptic Nucleus/pathology , Time Factors , Vasoconstrictor Agents/metabolism , Water/metabolism
18.
Mol Ther ; 5(5 Pt 1): 509-16, 2002 May.
Article in English | MEDLINE | ID: mdl-11991741

ABSTRACT

Viral vectors are excellent tools for studying gene function in the brain, although a limitation has been the ability to effectively target transgene expression to specific neuronal populations. This generally cannot be overcome by the use of neuron-specific promoters, as most are too large to be used with current viral vectors and expression from these promoters is often relatively weak. We therefore developed a composite expression cassette, comprising 495 bp of the weak human SYN1 (synapsin-1) promoter and 800 bp of the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE). Studies in hippocampal cultures, organotypic cultures, and in vivo showed that the 3' addition of the WPRE to the SYN1 element greatly increased enhanced green fluorescent protein expression levels with no loss of neuronal specificity. In vivo studies also showed that transgene expression was enhanced with no loss of neuronal specificity in dentate-gyrus neurons for at least 6 weeks following transfection. Therefore, unlike most powerful promoter systems, which mediate expression in neurons and glia, this SYN1-WPRE cassette can target powerful long-term transgene expression to central nervous system neurons when delivered at relatively low titers of adenovirus. Its use should therefore facilitate both gene therapy studies and investigations of neuronal gene function.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/administration & dosage , Hepatitis B Virus, Woodchuck/genetics , Hippocampus/metabolism , Luminescent Proteins/metabolism , Neurons/metabolism , Synapsins/genetics , Synapsins/metabolism , Animals , Cell Line , Flow Cytometry , Gene Expression Regulation, Viral , Gene Transfer Techniques , Genetic Vectors/genetics , Green Fluorescent Proteins , HeLa Cells , Humans , Immunoenzyme Techniques , Luminescent Proteins/genetics , Male , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , RNA, Viral/biosynthesis , RNA, Viral/genetics , Rats , Rats, Wistar , Regulatory Sequences, Nucleic Acid , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...